Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Cell ; 184(15): 3852-3872, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34297930

RESUMEN

Fibroblasts are diverse mesenchymal cells that participate in tissue homeostasis and disease by producing complex extracellular matrix and creating signaling niches through biophysical and biochemical cues. Transcriptionally and functionally heterogeneous across and within organs, fibroblasts encode regional positional information and maintain distinct cellular progeny. We summarize their development, lineages, functions, and contributions to fibrosis in four fibroblast-rich organs: skin, lung, skeletal muscle, and heart. We propose that fibroblasts are uniquely poised for tissue repair by easily reentering the cell cycle and exhibiting a reversible plasticity in phenotype and cell fate. These properties, when activated aberrantly, drive fibrotic disorders in humans.


Asunto(s)
Enfermedad , Fibroblastos/metabolismo , Salud , Animales , Linaje de la Célula , Humanos , Terapia Molecular Dirigida , Transducción de Señal
2.
J Immunol ; 212(8): 1287-1306, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38426910

RESUMEN

Myocarditis has emerged as an immune-related adverse event of immune checkpoint inhibitor (ICI) cancer therapy associated with significant mortality. To ensure patients continue to safely benefit from life-saving cancer therapy, an understanding of fundamental immunological phenomena underlying ICI myocarditis is essential. We recently developed the NOD-cMHCI/II-/-.DQ8 mouse model that spontaneously develops myocarditis with lower mortality than observed in previous HLA-DQ8 NOD mouse strains. Our strain was rendered murine MHC class I and II deficient using CRISPR/Cas9 technology, making it a genetically clean platform for dissecting CD4+ T cell-mediated myocarditis in the absence of classically selected CD8+ T cells. These mice are highly susceptible to myocarditis and acute heart failure following anti-PD-1 ICI-induced treatment. Additionally, anti-PD-1 administration accelerates skeletal muscle myositis. Using histology, flow cytometry, adoptive transfers, and RNA sequencing analyses, we performed a thorough characterization of cardiac and skeletal muscle T cells, identifying shared and unique characteristics of both populations. Taken together, this report details a mouse model with features of a rare, but highly lethal clinical presentation of overlapping myocarditis and myositis following ICI therapy. This study sheds light on underlying immunological mechanisms in ICI myocarditis and provides the basis for further detailed analyses of diagnostic and therapeutic strategies.


Asunto(s)
Diabetes Mellitus Experimental , Antígenos HLA-DQ , Miocarditis , Miositis , Neoplasias , Humanos , Ratones , Animales , Ratones Endogámicos NOD , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Miositis/inducido químicamente , Miositis/patología
3.
J Immunol ; 211(12): 1792-1805, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37877672

RESUMEN

In an effort to improve HLA-"humanized" mouse models for type 1 diabetes (T1D) therapy development, we previously generated directly in the NOD strain CRISPR/Cas9-mediated deletions of various combinations of murine MHC genes. These new models improved upon previously available platforms by retaining ß2-microglobulin functionality in FcRn and nonclassical MHC class I formation. As proof of concept, we generated H2-Db/H2-Kd double knockout NOD mice expressing human HLA-A*0201 or HLA-B*3906 class I variants that both supported autoreactive diabetogenic CD8+ T cell responses. In this follow-up work, we now describe the creation of 10 new NOD-based mouse models expressing various combinations of HLA genes with and without chimeric transgenic human TCRs reactive to proinsulin/insulin. The new TCR-transgenic models develop differing levels of insulitis mediated by HLA-DQ8-restricted insulin-reactive T cells. Additionally, these transgenic T cells can transfer insulitis to newly developed NSG mice lacking classical murine MHC molecules, but expressing HLA-DQ8. These new models can be used to test potential therapeutics for a possible capacity to reduce islet infiltration or change the phenotype of T cells expressing type 1 diabetes patient-derived ß cell autoantigen-specific TCRs.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Antígenos HLA-DQ , Humanos , Ratones , Animales , Ratones Endogámicos NOD , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/terapia , Insulina , Ratones Transgénicos , Ratones Noqueados , Receptores de Antígenos de Linfocitos T/genética
4.
J Mol Cell Cardiol ; 192: 48-64, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38734060

RESUMEN

INTRODUCTION: Chronic immunopathology contributes to the development of heart failure after a myocardial infarction. Both T and B cells of the adaptive immune system are present in the myocardium and have been suggested to be involved in post-MI immunopathology. METHODS: We analyzed the B and T cell populations isolated from previously published single cell RNA-sequencing data sets (PMID: 32130914, PMID: 35948637, PMID: 32971526 and PMID: 35926050), of the mouse and human heart, using differential expression analysis, functional enrichment analysis, gene regulatory inferences, and integration with autoimmune and cardiovascular GWAS. RESULTS: Already at baseline, mature effector B and T cells are present in the human and mouse heart, having increased activity in transcription factors maintaining tolerance (e.g. DEAF1, JDP2, SPI-B). Following MI, T cells upregulate pro-inflammatory transcript levels (e.g. Cd11, Gzmk, Prf1), while B cells upregulate activation markers (e.g. Il6, Il1rn, Ccl6) and collagen (e.g. Col5a2, Col4a1, Col1a2). Importantly, pro-inflammatory and fibrotic transcription factors (e.g. NFKB1, CREM, REL) remain active in T cells, while B cells maintain elevated activity in transcription factors related to immunoglobulin production (e.g. ERG, REL) in both mouse and human post-MI hearts. Notably, genes differentially expressed in post-MI T and B cells are associated with cardiovascular and autoimmune disease. CONCLUSION: These findings highlight the varied and time-dependent dynamic roles of post-MI T and B cells. They appear ready-to-go and are activated immediately after MI, thus participate in the acute wound healing response. However, they subsequently remain in a state of pro-inflammatory activation contributing to persistent immunopathology.


Asunto(s)
Linfocitos B , Infarto del Miocardio , Miocardio , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Infarto del Miocardio/genética , Infarto del Miocardio/inmunología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Humanos , Animales , Ratones , Miocardio/metabolismo , Miocardio/patología , Linfocitos B/metabolismo , Linfocitos B/inmunología , Linfocitos T/metabolismo , Linfocitos T/inmunología , Inmunidad Adaptativa/genética , Regulación de la Expresión Génica , Perfilación de la Expresión Génica , Transcriptoma/genética , Transcripción Genética , Estudio de Asociación del Genoma Completo
5.
Circulation ; 143(8): 821-836, 2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33297741

RESUMEN

BACKGROUND: Ischemic heart disease is a leading cause of heart failure and despite advanced therapeutic options, morbidity and mortality rates remain high. Although acute inflammation in response to myocardial cell death has been extensively studied, subsequent adaptive immune activity and anti-heart autoimmunity may also contribute to the development of heart failure. After ischemic injury to the myocardium, dendritic cells (DC) respond to cardiomyocyte necrosis, present cardiac antigen to T cells, and potentially initiate a persistent autoimmune response against the heart. Cross-priming DC have the ability to activate both CD4+ helper and CD8+ cytotoxic T cells in response to necrotic cells and may thus be crucial players in exacerbating autoimmunity targeting the heart. This study investigates a role for cross-priming DC in post-myocardial infarction immunopathology through presentation of self-antigen from necrotic cardiac cells to cytotoxic CD8+ T cells. METHODS: We induced type 2 myocardial infarction-like ischemic injury in the heart by treatment with a single high dose of the ß-adrenergic agonist isoproterenol. We characterized the DC population in the heart and mediastinal lymph nodes and analyzed long-term cardiac immunopathology and functional decline in wild type and Clec9a-depleted mice lacking DC cross-priming function. RESULTS: A diverse DC population, including cross-priming DC, is present in the heart and activated after ischemic injury. Clec9a-/- mice deficient in DC cross-priming are protected from persistent immune-mediated myocardial damage and decline of cardiac function, likely because of dampened activation of cytotoxic CD8+ T cells. CONCLUSION: Activation of cytotoxic CD8+ T cells by cross-priming DC contributes to exacerbation of postischemic inflammatory damage of the myocardium and corresponding decline in cardiac function. Importantly, this provides novel therapeutic targets to prevent postischemic immunopathology and heart failure.


Asunto(s)
Reactividad Cruzada , Células Dendríticas/inmunología , Miocardio/patología , Animales , Presentación de Antígeno , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Insuficiencia Cardíaca/patología , Humanos , Lectinas Tipo C/deficiencia , Lectinas Tipo C/genética , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Infarto del Miocardio/inmunología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/inmunología , Miocardio/metabolismo , Receptores de Quimiocina/metabolismo , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética
6.
J Cell Mol Med ; 25(1): 229-243, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33249764

RESUMEN

Heart failure is the common final pathway of several cardiovascular conditions and a major cause of morbidity and mortality worldwide. Aberrant activation of the adaptive immune system in response to myocardial necrosis has recently been implicated in the development of heart failure. The ß-adrenergic agonist isoproterenol hydrochloride is used for its cardiac effects in a variety of different dosing regimens with high doses causing acute cardiomyocyte necrosis. To assess whether isoproterenol-induced cardiomyocyte necrosis triggers an adaptive immune response against the heart, we treated C57BL/6J mice with a single intraperitoneal injection of isoproterenol. We confirmed tissue damage reminiscent of human type 2 myocardial infarction. This is followed by an adaptive immune response targeting the heart as demonstrated by the activation of T cells, the presence of anti-heart auto-antibodies in the serum as late as 12 weeks after initial challenge and IgG deposition in the myocardium. All of these are hallmark signs of an established autoimmune response. Adoptive transfer of splenocytes from isoproterenol-treated mice induces left ventricular dilation and impairs cardiac function in healthy recipients. In summary, a single administration of a high dose of isoproterenol is a suitable high-throughput model for future studies of the pathological mechanisms of anti-heart autoimmunity and to test potential immunomodulatory therapeutic approaches.


Asunto(s)
Inmunidad Adaptativa , Infarto del Miocardio/inmunología , Miocardio/patología , Traslado Adoptivo , Animales , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Femenino , Fibrosis , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Isoproterenol , Antígenos Comunes de Leucocito/metabolismo , Masculino , Ratones Endogámicos C57BL , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Necrosis , Especificidad de Órganos , Bazo/inmunología , Sístole , Linfocitos T Colaboradores-Inductores/inmunología , Vasodilatación
7.
Int J Mol Sci ; 22(4)2021 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-33669808

RESUMEN

Recent technological advances have revolutionized the study of tissue biology and garnered a greater appreciation for tissue complexity. In order to understand cardiac development, heart tissue homeostasis, and the effects of stress and injury on the cardiovascular system, it is essential to characterize the heart at high cellular resolution. Single-cell profiling provides a more precise definition of tissue composition, cell differentiation trajectories, and intercellular communication, compared to classical bulk approaches. Here, we aim to review how recent single-cell multi-omic studies have changed our understanding of cell dynamics during cardiac development, and in the healthy and diseased adult myocardium.


Asunto(s)
Sistema Cardiovascular/citología , Análisis de la Célula Individual , Transcriptoma/genética , Animales , COVID-19/genética , COVID-19/patología , Reprogramación Celular/genética , Desarrollo Embrionario/genética , Humanos
8.
Development ; 143(8): 1242-58, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-27095490

RESUMEN

Over the past two decades, several populations of cardiac stem cells have been described in the adult mammalian heart. For the most part, however, their lineage origins and in vivo functions remain largely unexplored. This Review summarizes what is known about different populations of embryonic and adult cardiac stem cells, including KIT(+), PDGFRα(+), ISL1(+)and SCA1(+)cells, side population cells, cardiospheres and epicardial cells. We discuss their developmental origins and defining characteristics, and consider their possible contribution to heart organogenesis and regeneration. We also summarize the origin and plasticity of cardiac fibroblasts and circulating endothelial progenitor cells, and consider what role these cells have in contributing to cardiac repair.


Asunto(s)
Linaje de la Célula , Miocardio/citología , Células Madre/citología , Células Madre Adultas , Animales , Diferenciación Celular , Células Madre Embrionarias , Humanos
10.
J Cell Mol Med ; 18(4): 624-34, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24444305

RESUMEN

Cardiac progenitor cells (CPCs) isolated as cardiospheres (CSs) and CS-derived cells (CDCs) are a promising tool for cardiac cell therapy in heart failure patients, having CDCs already been used in a phase I/II clinical trial. Culture standardization according to Good Manufacturing Practices (GMPs) is a mandatory step for clinical translation. One of the main issues raised is the use of xenogenic additives (e.g. FBS, foetal bovine serum) in cell culture media, which carries the risk of contamination with infectious viral/prion agents, and the possible induction of immunizing effects in the final recipient. In this study, B27 supplement and sera requirements to comply with European GMPs were investigated in CSs and CDCs cultures, in terms of process yield/efficiency and final cell product gene expression levels, as well as phenotype. B27- free CS cultures produced a significantly reduced yield and a 10-fold drop in c-kit expression levels versus B27+ media. Moreover, autologous human serum (aHS) and two different commercially available GMP AB HSs were compared with standard research-grade FBS. CPCs from all HSs explants had reduced growth rate, assumed a senescent-like morphology with time in culture, and/or displayed a significant shift towards the endothelial phenotype. Among three different GMP gamma-irradiated FBSs (giFBSs) tested, two provided unsatisfactory cell yields, while one performed optimally, in terms of CPCs yield/phenotype. In conclusion, the use of HSs for the isolation and expansion of CSs/CDCs has to be excluded because of altered proliferation and/or commitment, while media supplemented with B27 and the selected giFBS allows successful EU GMP-complying CPCs culture.


Asunto(s)
Técnicas de Cultivo de Célula , Medios de Cultivo/química , Suero/química , Células Madre/citología , Animales , Bovinos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas Proto-Oncogénicas c-kit/biosíntesis , Células Madre/efectos de los fármacos
11.
Biochim Biophys Acta ; 1830(2): 2459-69, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22921810

RESUMEN

BACKGROUND: Cardiac regenerative medicine is a rapidly evolving field, with promising future developments for effective personalized treatments. Several stem/progenitor cells are candidates for cardiac cell therapy, and emerging evidence suggests how multiple metabolic and biochemical pathways strictly regulate their fate and renewal. SCOPE OF REVIEW: In this review, we will explore a selection of areas of common interest for biology and biochemistry concerning stem/progenitor cells, and in particular cardiac progenitor cells. Numerous regulatory mechanisms have been identified that link stem cell signaling and functions to the modulation of metabolic pathways, and vice versa. Pharmacological treatments and culture requirements may be exploited to modulate stem cell pluripotency and self-renewal, possibly boosting their regenerative potential for cell therapy. MAJOR CONCLUSIONS: Mitochondria and their many related metabolites and messengers, such as oxygen, ROS, calcium and glucose, have a crucial role in regulating stem cell fate and the balance of their functions, together with many metabolic enzymes. Furthermore, protein biochemistry and proteomics can provide precious clues on the definition of different progenitor cell populations, their physiology and their autocrine/paracrine regulatory/signaling networks. GENERAL SIGNIFICANCE: Interdisciplinary approaches between biology and biochemistry can provide productive insights on stem/progenitor cells, allowing the development of novel strategies and protocols for effective cardiac cell therapy clinical translation. This article is part of a Special Issue entitled Biochemistry of Stem Cells.


Asunto(s)
Miocardio/citología , Células Madre/citología , Humanos
12.
bioRxiv ; 2023 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-35233576

RESUMEN

Inflammation in response to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection drives severity of coronavirus disease 2019 (COVID-19) and is influenced by host genetics. To understand mechanisms of inflammation, animal models that reflect genetic diversity and clinical outcomes observed in humans are needed. We report a mouse panel comprising the genetically diverse Collaborative Cross (CC) founder strains crossed to human ACE2 transgenic mice (K18-hACE2) that confers susceptibility to SARS-CoV-2. Infection of CC x K18- hACE2 resulted in a spectrum of survival, viral replication kinetics, and immune profiles. Importantly, in contrast to the K18-hACE2 model, early type I interferon (IFN-I) and regulated proinflammatory responses were required for control of SARS-CoV-2 replication in PWK x K18-hACE2 mice that were highly resistant to disease. Thus, virus dynamics and inflammation observed in COVID-19 can be modeled in diverse mouse strains that provide a genetically tractable platform for understanding anti-coronavirus immunity.

13.
Nat Commun ; 14(1): 4481, 2023 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-37491352

RESUMEN

Inflammation in response to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection drives severity of coronavirus disease 2019 (COVID-19) and is influenced by host genetics. To understand mechanisms of inflammation, animal models that reflect genetic diversity and clinical outcomes observed in humans are needed. We report a mouse panel comprising the genetically diverse Collaborative Cross (CC) founder strains crossed to human ACE2 transgenic mice (K18-hACE2) that confers susceptibility to SARS-CoV-2. Infection of CC x K18-hACE2 resulted in a spectrum of survival, viral replication kinetics, and immune profiles. Importantly, in contrast to the K18-hACE2 model, early type I interferon (IFN-I) and regulated proinflammatory responses were required for control of SARS-CoV-2 replication in PWK x K18-hACE2 mice that were highly resistant to disease. Thus, virus dynamics and inflammation observed in COVID-19 can be modeled in diverse mouse strains that provide a genetically tractable platform for understanding anti-coronavirus immunity.


Asunto(s)
COVID-19 , Interferón Tipo I , Humanos , Ratones , Animales , Citocinas , SARS-CoV-2 , Ratones Transgénicos , Inflamación/genética , Modelos Animales de Enfermedad , Pulmón
14.
Elife ; 112022 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-35293863

RESUMEN

Organ fibroblasts are essential components of homeostatic and diseased tissues. They participate in sculpting the extracellular matrix, sensing the microenvironment, and communicating with other resident cells. Recent studies have revealed transcriptomic heterogeneity among fibroblasts within and between organs. To dissect the basis of interorgan heterogeneity, we compare the gene expression of murine fibroblasts from different tissues (tail, skin, lung, liver, heart, kidney, and gonads) and show that they display distinct positional and organ-specific transcriptome signatures that reflect their embryonic origins. We demonstrate that expression of genes typically attributed to the surrounding parenchyma by fibroblasts is established in embryonic development and largely maintained in culture, bioengineered tissues and ectopic transplants. Targeted knockdown of key organ-specific transcription factors affects fibroblast functions, in particular genes involved in the modulation of fibrosis and inflammation. In conclusion, our data reveal that adult fibroblasts maintain an embryonic gene expression signature inherited from their organ of origin, thereby increasing our understanding of adult fibroblast heterogeneity. The knowledge of this tissue-specific gene signature may assist in targeting fibrotic diseases in a more precise, organ-specific manner.


Asunto(s)
Fibroblastos , Transcriptoma , Animales , Fibroblastos/metabolismo , Fibrosis , Pulmón/metabolismo , Ratones , Piel/metabolismo
15.
J Cell Mol Med ; 15(1): 63-71, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19912439

RESUMEN

Experimental data suggest that cell-based therapies may be useful for cardiac regeneration following ischaemic heart disease. Bone marrow (BM) cells have been reported to contribute to tissue repair after myocardial infarction (MI) by a variety of humoural and cellular mechanisms. However, there is no direct evidence, so far, that BM cells can generate cardiac stem cells (CSCs). To investigate whether BM cells contribute to repopulate the Kit(+) CSCs pool, we transplanted BM cells from transgenic mice, expressing green fluorescent protein under the control of Kit regulatory elements, into wild-type irradiated recipients. Following haematological reconstitution and MI, CSCs were cultured from cardiac explants to generate 'cardiospheres', a microtissue normally originating in vitro from CSCs. These were all green fluorescent (i.e. BM derived) and contained cells capable of initiating differentiation into cells expressing the cardiac marker Nkx2.5. These findings indicate that, at least in conditions of local acute cardiac damage, BM cells can home into the heart and give rise to cells that share properties of resident Kit(+) CSCs.


Asunto(s)
Células de la Médula Ósea/citología , Diferenciación Celular , Cardiopatías/cirugía , Miocitos Cardíacos/fisiología , Miocitos Cardíacos/trasplante , Células Madre/fisiología , Animales , Animales Recién Nacidos , Western Blotting , Células de la Médula Ósea/metabolismo , Femenino , Cardiopatías/patología , Ratones , Ratones Transgénicos , ARN Mensajero/genética , Regeneración , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Nat Commun ; 12(1): 681, 2021 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-33514719

RESUMEN

Endothelial cells play a critical role in the adaptation of tissues to injury. Tissue ischemia induced by infarction leads to profound changes in endothelial cell functions and can induce transition to a mesenchymal state. Here we explore the kinetics and individual cellular responses of endothelial cells after myocardial infarction by using single cell RNA sequencing. This study demonstrates a time dependent switch in endothelial cell proliferation and inflammation associated with transient changes in metabolic gene signatures. Trajectory analysis reveals that the majority of endothelial cells 3 to 7 days after myocardial infarction acquire a transient state, characterized by mesenchymal gene expression, which returns to baseline 14 days after injury. Lineage tracing, using the Cdh5-CreERT2;mT/mG mice followed by single cell RNA sequencing, confirms the transient mesenchymal transition and reveals additional hypoxic and inflammatory signatures of endothelial cells during early and late states after injury. These data suggest that endothelial cells undergo a transient mes-enchymal activation concomitant with a metabolic adaptation within the first days after myocardial infarction but do not acquire a long-term mesenchymal fate. This mesenchymal activation may facilitate endothelial cell migration and clonal expansion to regenerate the vascular network.


Asunto(s)
Endotelio/patología , Transición Epitelial-Mesenquimal/genética , Infarto del Miocardio/patología , Miocardio/patología , Animales , Movimiento Celular/genética , Plasticidad de la Célula/genética , Proliferación Celular/genética , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/patología , Endotelio/citología , Genes Reporteros/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Proteínas Luminiscentes/genética , Masculino , Ratones , Ratones Transgénicos , Miocardio/citología , RNA-Seq , Análisis de la Célula Individual
17.
J Cell Mol Med ; 14(5): 1071-7, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20465579

RESUMEN

Tissue engineering is an increasingly expanding area of research in the cardiovascular field that involves engineering, chemistry, biology and medicine. Cardiac tissue engineering (CTE) aims to regenerate myocardial damage by combining cells, matrix, biological active molecules and physiological stimuli. The rationale behind CTE applications is that in order to regenerate the ventricular wall after a myocardial infarction it is necessary to combine procedures that regenerate both cardiomyocytes and the extracellular matrix. The application of (stem) cells together with a matrix could represent an environment protected from the inflammatory and pro-apoptotic signals, a stemness/survival reservoir slowly releasing cells and factors promoting tissue regeneration and angiogenesis. This review will focus on the applications and advantages that CTE application could offer compared to conventional cell therapy.


Asunto(s)
Miocardio/citología , Miocardio/metabolismo , Regeneración/fisiología , Esferoides Celulares/citología , Ingeniería de Tejidos/métodos , Humanos , Esferoides Celulares/metabolismo , Trasplante de Células Madre
18.
STAR Protoc ; 1(2): 100077, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-33000003

RESUMEN

Interstitial cells have a crucial role in cardiac fibrosis and repair of the mammalian heart. Single-cell profiling using droplet-based technology has revolutionized the investigation of cell states and identities. Here, we present a protocol for the efficient isolation of high-quality live nucleated non-cardiomyocytes from adult murine heart, for unbiased single-cell RNA sequencing using 10× Chromium technology. This protocol has been applied to homeostatic and injured hearts from different mouse strains. For complete details on the use and execution of this protocol, please refer to Forte et al. (2020).


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Cultivadas/citología , Miocardio/citología , Análisis de la Célula Individual/métodos , Animales , Ratones
19.
Cell Rep ; 30(9): 3149-3163.e6, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32130914

RESUMEN

Cardiac ischemia leads to the loss of myocardial tissue and the activation of a repair process that culminates in the formation of a scar whose structural characteristics dictate propensity to favorable healing or detrimental cardiac wall rupture. To elucidate the cellular processes underlying scar formation, here we perform unbiased single-cell mRNA sequencing of interstitial cells isolated from infarcted mouse hearts carrying a genetic tracer that labels epicardial-derived cells. Sixteen interstitial cell clusters are revealed, five of which were of epicardial origin. Focusing on stromal cells, we define 11 sub-clusters, including diverse cell states of epicardial- and endocardial-derived fibroblasts. Comparing transcript profiles from post-infarction hearts in C57BL/6J and 129S1/SvImJ inbred mice, which displays a marked divergence in the frequency of cardiac rupture, uncovers an early increase in activated myofibroblasts, enhanced collagen deposition, and persistent acute phase response in 129S1/SvImJ mouse hearts, defining a crucial time window of pathological remodeling that predicts disease outcome.


Asunto(s)
Infarto del Miocardio/genética , Miocardio/patología , Rotura/patología , Animales , Cicatriz/patología , Homeostasis , Ratones , Ratones Endogámicos , Miofibroblastos/patología , Pericardio/patología , Fenotipo , RNA-Seq , Análisis de la Célula Individual , Células del Estroma/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA